Skip to main content
Erschienen in: Endocrine 1/2015

01.09.2015 | Original Article

Autocrine effect of Zn2+ on the glucose-stimulated insulin secretion

verfasst von: Kira G. Slepchenko, Nigel A. Daniels, Aili Guo, Yang V. Li

Erschienen in: Endocrine | Ausgabe 1/2015

Einloggen, um Zugang zu erhalten

Abstract

It is well known that zinc (Zn2+) is required for the process of insulin biosynthesis and the maturation of insulin secretory granules in pancreatic beta (β)-cells, and that changes in Zn2+ levels in the pancreas have been found to be associated with diabetes. Glucose-stimulation causes a rapid co-secretion of Zn2+ and insulin with similar kinetics. However, we do not know whether Zn2+ regulates insulin availability and secretion. Here we investigated the effect of Zn2+ on glucose-stimulated insulin secretion (GSIS) in isolated mouse pancreatic islets. Whereas Zn2+ alone (control) had no effect on the basal secretion of insulin, it significantly inhibited GSIS. The application of CaEDTA, by removing the secreted Zn2+ from the extracellular milieu of the islets, resulted in significantly increased GSIS, suggesting an overall inhibitory role of secreted Zn2+ on GSIS. The inhibitory action of Zn2+ was mostly mediated through the activities of KATP/Ca2+ channels. Furthermore, during brief paired-pulse glucose-stimulated Zn2+ secretion (GSZS), Zn2+ secretion following the second pulse was significantly attenuated, probably by the secreted endogenous Zn2+ after the first pulse. Such an inhibition on Zn2+ secretion following the second pulse was completely reversed by Zn2+ chelation, suggesting a negative feedback mechanism, in which the initial glucose-stimulated Zn2+ release inhibits subsequent Zn2+ secretion, subsequently inhibiting insulin co-secretion as well. Taken together, these data suggest a negative feedback mechanism on GSZS and GSIS by Zn2+ secreted from β-cells, and the co-secreted Zn2+ may act as an autocrine inhibitory modulator.
Literatur
3.
4.
Zurück zum Zitat K. Lemaire, M.A. Ravier, A. Schraenen, J.W. Creemers, R. Van de Plas, M. Granvik, L. Van Lommel, E. Waelkens, F. Chimienti, G.A. Rutter, P. Gilon, P.A. in’t Veld, F.C. Schuit, Insulin crystallization depends on zinc transporter ZnT8 expression, but is not required for normal glucose homeostasis in mice. Proc Natl Acad Sci U S A 106(35), 14872–14877 (2009). doi:10.1073/pnas.0906587106 PubMedCentralCrossRefPubMed K. Lemaire, M.A. Ravier, A. Schraenen, J.W. Creemers, R. Van de Plas, M. Granvik, L. Van Lommel, E. Waelkens, F. Chimienti, G.A. Rutter, P. Gilon, P.A. in’t Veld, F.C. Schuit, Insulin crystallization depends on zinc transporter ZnT8 expression, but is not required for normal glucose homeostasis in mice. Proc Natl Acad Sci U S A 106(35), 14872–14877 (2009). doi:10.​1073/​pnas.​0906587106 PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat J. Brandao-Neto, C.A. Silva, T. Shuhama, J.A. Silva, L. Oba, Renal handling of zinc in insulin-dependent diabetes mellitus patients. Biometals 14(1), 75–80 (2001)CrossRefPubMed J. Brandao-Neto, C.A. Silva, T. Shuhama, J.A. Silva, L. Oba, Renal handling of zinc in insulin-dependent diabetes mellitus patients. Biometals 14(1), 75–80 (2001)CrossRefPubMed
6.
Zurück zum Zitat J.J. Cunningham, A. Fu, P.L. Mearkle, R.G. Brown, Hyperzincuria in individuals with insulin-dependent diabetes mellitus: concurrent zinc status and the effect of high-dose zinc supplementation. Metabolism 43(12), 1558–1562 (1994)CrossRefPubMed J.J. Cunningham, A. Fu, P.L. Mearkle, R.G. Brown, Hyperzincuria in individuals with insulin-dependent diabetes mellitus: concurrent zinc status and the effect of high-dose zinc supplementation. Metabolism 43(12), 1558–1562 (1994)CrossRefPubMed
8.
Zurück zum Zitat P. Proks, J.D. Lippiat, Membrane ion channels and diabetes. Curr. Pharm. Des. 12(4), 485–501 (2006)CrossRefPubMed P. Proks, J.D. Lippiat, Membrane ion channels and diabetes. Curr. Pharm. Des. 12(4), 485–501 (2006)CrossRefPubMed
13.
Zurück zum Zitat Y.V. Li, Zinc Overload in Stroke, in Metal Ion in Stroke, ed. by Y.V. Li, J.H. Zhang (Springer Science+Business Media, New York, 2012), pp. 167–189CrossRef Y.V. Li, Zinc Overload in Stroke, in Metal Ion in Stroke, ed. by Y.V. Li, J.H. Zhang (Springer Science+Business Media, New York, 2012), pp. 167–189CrossRef
14.
Zurück zum Zitat Y.V. Li, C.J. Hough, J.M. Sarvey, Do we need zinc to think? Sci. STKE 182, 19 (2003) Y.V. Li, C.J. Hough, J.M. Sarvey, Do we need zinc to think? Sci. STKE 182, 19 (2003)
17.
Zurück zum Zitat A. Mathie, G.L. Sutton, C.E. Clarke, E.L. Veale, Zinc and copper: pharmacological probes and endogenous modulators of neuronal excitability. Pharmacol. Ther. 111(3), 567–583 (2006)CrossRefPubMed A. Mathie, G.L. Sutton, C.E. Clarke, E.L. Veale, Zinc and copper: pharmacological probes and endogenous modulators of neuronal excitability. Pharmacol. Ther. 111(3), 567–583 (2006)CrossRefPubMed
18.
Zurück zum Zitat D.W. Barnett, D.M. Pressel, S. Misler, Voltage-dependent Na+ and Ca2+ currents in human pancreatic islet beta-cells: evidence for roles in the generation of action potentials and insulin secretion. Pflugers Arch. 431(2), 272–282 (1995)CrossRefPubMed D.W. Barnett, D.M. Pressel, S. Misler, Voltage-dependent Na+ and Ca2+ currents in human pancreatic islet beta-cells: evidence for roles in the generation of action potentials and insulin secretion. Pflugers Arch. 431(2), 272–282 (1995)CrossRefPubMed
20.
Zurück zum Zitat A. Tarasov, J. Dusonchet, F. Ashcroft, Metabolic regulation of the pancreatic beta-cell ATP-sensitive K+ channel: a pas de deux. Diabetes 53(Suppl 3), S113–S122 (2004)CrossRefPubMed A. Tarasov, J. Dusonchet, F. Ashcroft, Metabolic regulation of the pancreatic beta-cell ATP-sensitive K+ channel: a pas de deux. Diabetes 53(Suppl 3), S113–S122 (2004)CrossRefPubMed
21.
Zurück zum Zitat F.M. Ashcroft, P. Rorsman, G. Trube, Single calcium channel activity in mouse pancreatic beta-cells. Ann. N. Y. Acad. Sci. 560, 410–412 (1989)CrossRefPubMed F.M. Ashcroft, P. Rorsman, G. Trube, Single calcium channel activity in mouse pancreatic beta-cells. Ann. N. Y. Acad. Sci. 560, 410–412 (1989)CrossRefPubMed
22.
Zurück zum Zitat I. Findlay, F.M. Ashcroft, R.P. Kelly, P. Rorsman, O.H. Petersen, G. Trube, Calcium currents in insulin-secreting beta-cells. Ann. N. Y. Acad. Sci. 560, 403–409 (1989)CrossRefPubMed I. Findlay, F.M. Ashcroft, R.P. Kelly, P. Rorsman, O.H. Petersen, G. Trube, Calcium currents in insulin-secreting beta-cells. Ann. N. Y. Acad. Sci. 560, 403–409 (1989)CrossRefPubMed
23.
Zurück zum Zitat A. Bloc, T. Cens, H. Cruz, Y. Dunant, Zinc-induced changes in ionic currents of clonal rat pancreatic -cells: activation of ATP-sensitive K+ channels. J. Physiol. 529(Pt 3), 723–734 (2000)PubMedCentralCrossRefPubMed A. Bloc, T. Cens, H. Cruz, Y. Dunant, Zinc-induced changes in ionic currents of clonal rat pancreatic -cells: activation of ATP-sensitive K+ channels. J. Physiol. 529(Pt 3), 723–734 (2000)PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat R. Ferrer, B. Soria, C.M. Dawson, I. Atwater, E. Rojas, Effects of Zn2+ on glucose-induced electrical activity and insulin release from mouse pancreatic islets. Am. J. Physiol. 246(5 Pt 1), C520–C527 (1984)PubMed R. Ferrer, B. Soria, C.M. Dawson, I. Atwater, E. Rojas, Effects of Zn2+ on glucose-induced electrical activity and insulin release from mouse pancreatic islets. Am. J. Physiol. 246(5 Pt 1), C520–C527 (1984)PubMed
25.
Zurück zum Zitat T. Ghafghazi, M.L. McDaniel, P.E. Lacy, Zinc-induced inhibition of insulin secretion from isolated rat islets of Langerhans. Diabetes 30(4), 341–345 (1981)CrossRefPubMed T. Ghafghazi, M.L. McDaniel, P.E. Lacy, Zinc-induced inhibition of insulin secretion from isolated rat islets of Langerhans. Diabetes 30(4), 341–345 (1981)CrossRefPubMed
27.
Zurück zum Zitat B. Holst, K.L. Egerod, C. Jin, P.S. Petersen, M.V. Ostergaard, J. Hald, A.M. Sprinkel, J. Storling, T. Mandrup-Poulsen, J.J. Holst, P. Thams, C. Orskov, N. Wierup, F. Sundler, O.D. Madsen, T.W. Schwartz, G protein-coupled receptor 39 deficiency is associated with pancreatic islet dysfunction. Endocrinology 150(6), 2577–2585 (2009). doi:10.1210/en.2008-1250 PubMedCentralCrossRefPubMed B. Holst, K.L. Egerod, C. Jin, P.S. Petersen, M.V. Ostergaard, J. Hald, A.M. Sprinkel, J. Storling, T. Mandrup-Poulsen, J.J. Holst, P. Thams, C. Orskov, N. Wierup, F. Sundler, O.D. Madsen, T.W. Schwartz, G protein-coupled receptor 39 deficiency is associated with pancreatic islet dysfunction. Endocrinology 150(6), 2577–2585 (2009). doi:10.​1210/​en.​2008-1250 PubMedCentralCrossRefPubMed
28.
29.
Zurück zum Zitat C.A. Aspinwall, S.A. Brooks, R.T. Kennedy, J.R. Lakey, Effects of intravesicular H+ and extracellular H+ and Zn2+ on insulin secretion in pancreatic beta cells. J. Biol. Chem. 272(50), 31308–31314 (1997)CrossRefPubMed C.A. Aspinwall, S.A. Brooks, R.T. Kennedy, J.R. Lakey, Effects of intravesicular H+ and extracellular H+ and Zn2+ on insulin secretion in pancreatic beta cells. J. Biol. Chem. 272(50), 31308–31314 (1997)CrossRefPubMed
30.
Zurück zum Zitat B. Ahren, Type 2 diabetes, insulin secretion and beta-cell mass. Curr. Mol. Med. 5(3), 275–286 (2005)CrossRefPubMed B. Ahren, Type 2 diabetes, insulin secretion and beta-cell mass. Curr. Mol. Med. 5(3), 275–286 (2005)CrossRefPubMed
31.
Zurück zum Zitat D.M. Muoio, C.B. Newgard, Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat. Rev. Mol. Cell Biol. 9(3), 193–205 (2008). doi:10.1038/nrm2327 CrossRefPubMed D.M. Muoio, C.B. Newgard, Mechanisms of disease: molecular and metabolic mechanisms of insulin resistance and beta-cell failure in type 2 diabetes. Nat. Rev. Mol. Cell Biol. 9(3), 193–205 (2008). doi:10.​1038/​nrm2327 CrossRefPubMed
34.
Zurück zum Zitat P.D. Zalewski, S.H. Millard, I.J. Forbes, O. Kapaniris, A. Slavotinek, W.H. Betts, A.D. Ward, S.F. Lincoln, I. Mahadevan, Video image analysis of labile zinc in viable pancreatic islet cells using a specific fluorescent probe for zinc. J. Histochem. Cytochem. 42(7), 877–884 (1994)CrossRefPubMed P.D. Zalewski, S.H. Millard, I.J. Forbes, O. Kapaniris, A. Slavotinek, W.H. Betts, A.D. Ward, S.F. Lincoln, I. Mahadevan, Video image analysis of labile zinc in viable pancreatic islet cells using a specific fluorescent probe for zinc. J. Histochem. Cytochem. 42(7), 877–884 (1994)CrossRefPubMed
35.
Zurück zum Zitat W.J. Qian, K.R. Gee, R.T. Kennedy, Imaging of Zn2 + release from pancreatic beta-cells at the level of single exocytotic events. Anal. Chem. 75(14), 3468–3475 (2003)CrossRefPubMed W.J. Qian, K.R. Gee, R.T. Kennedy, Imaging of Zn2 + release from pancreatic beta-cells at the level of single exocytotic events. Anal. Chem. 75(14), 3468–3475 (2003)CrossRefPubMed
36.
Zurück zum Zitat W.J. Qian, J.L. Peters, G.M. Dahlgren, K.R. Gee, R.T. Kennedy, Simultaneous monitoring of Zn2+ secretion and intracellular Ca2+ from islets and islet cells by fluorescence microscopy. Biotechniques 37(6), 922–924 (2004). 926, 928-930 passimPubMed W.J. Qian, J.L. Peters, G.M. Dahlgren, K.R. Gee, R.T. Kennedy, Simultaneous monitoring of Zn2+ secretion and intracellular Ca2+ from islets and islet cells by fluorescence microscopy. Biotechniques 37(6), 922–924 (2004). 926, 928-930 passimPubMed
37.
Zurück zum Zitat F. Chimienti, S. Devergnas, F. Pattou, F. Schuit, R. Garcia-Cuenca, B. Vandewalle, J. Kerr-Conte, L. Van Lommel, D. Grunwald, A. Favier, M. Seve, In vivo expression and functional characterization of the zinc transporter ZnT8 in glucose-induced insulin secretion. J. Cell Sci. 119(Pt 20), 4199–4206 (2006). doi:10.1242/jcs.03164 CrossRefPubMed F. Chimienti, S. Devergnas, F. Pattou, F. Schuit, R. Garcia-Cuenca, B. Vandewalle, J. Kerr-Conte, L. Van Lommel, D. Grunwald, A. Favier, M. Seve, In vivo expression and functional characterization of the zinc transporter ZnT8 in glucose-induced insulin secretion. J. Cell Sci. 119(Pt 20), 4199–4206 (2006). doi:10.​1242/​jcs.​03164 CrossRefPubMed
38.
Zurück zum Zitat B. Formby, F. Schmid-Formby, G.M. Grodsky, Relationship between insulin release and 65zinc efflux from rat pancreatic islets maintained in tissue culture. Diabetes 33(3), 229–234 (1984)CrossRefPubMed B. Formby, F. Schmid-Formby, G.M. Grodsky, Relationship between insulin release and 65zinc efflux from rat pancreatic islets maintained in tissue culture. Diabetes 33(3), 229–234 (1984)CrossRefPubMed
40.
Zurück zum Zitat D.P. Figlewicz, B. Formby, A.T. Hodgson, F.G. Schmid, G.M. Grodsky, Kinetics of 65zinc uptake and distribution in fractions from cultured rat islets of langerhans. Diabetes 29(10), 767–773 (1980)CrossRefPubMed D.P. Figlewicz, B. Formby, A.T. Hodgson, F.G. Schmid, G.M. Grodsky, Kinetics of 65zinc uptake and distribution in fractions from cultured rat islets of langerhans. Diabetes 29(10), 767–773 (1980)CrossRefPubMed
41.
Zurück zum Zitat G. Gold, G.M. Grodsky, Kinetic aspects of compartmental storage and secretion of insulin and zinc. Experientia 40(10), 1105–1114 (1984)CrossRefPubMed G. Gold, G.M. Grodsky, Kinetic aspects of compartmental storage and secretion of insulin and zinc. Experientia 40(10), 1105–1114 (1984)CrossRefPubMed
42.
Zurück zum Zitat E.D. Kilpatrick, R.P. Robertson, Differentiation between glucose-induced desensitization of insulin secretion and beta-cell exhaustion in the HIT-T15 cell line. Diabetes 47(4), 606–611 (1998)CrossRefPubMed E.D. Kilpatrick, R.P. Robertson, Differentiation between glucose-induced desensitization of insulin secretion and beta-cell exhaustion in the HIT-T15 cell line. Diabetes 47(4), 606–611 (1998)CrossRefPubMed
43.
Zurück zum Zitat R.F. Santerre, R.A. Cook, R.M. Crisel, J.D. Sharp, R.J. Schmidt, D.C. Williams, C.P. Wilson, Insulin synthesis in a clonal cell line of simian virus 40-transformed hamster pancreatic beta cells. Proc. Natl. Acad. Sci. U.S.A. 78(7), 4339–4343 (1981)PubMedCentralCrossRefPubMed R.F. Santerre, R.A. Cook, R.M. Crisel, J.D. Sharp, R.J. Schmidt, D.C. Williams, C.P. Wilson, Insulin synthesis in a clonal cell line of simian virus 40-transformed hamster pancreatic beta cells. Proc. Natl. Acad. Sci. U.S.A. 78(7), 4339–4343 (1981)PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat M. Skelin, M. Rupnik, A. Cencic, Pancreatic beta cell lines and their applications in diabetes mellitus research. Altex 27(2), 105–113 (2010)PubMed M. Skelin, M. Rupnik, A. Cencic, Pancreatic beta cell lines and their applications in diabetes mellitus research. Altex 27(2), 105–113 (2010)PubMed
45.
Zurück zum Zitat J. Miyazaki, K. Araki, E. Yamato, H. Ikegami, T. Asano, Y. Shibasaki, Y. Oka, K. Yamamura, Establishment of a pancreatic beta cell line that retains glucose-inducible insulin secretion: special reference to expression of glucose transporter isoforms. Endocrinology 127(1), 126–132 (1990)CrossRefPubMed J. Miyazaki, K. Araki, E. Yamato, H. Ikegami, T. Asano, Y. Shibasaki, Y. Oka, K. Yamamura, Establishment of a pancreatic beta cell line that retains glucose-inducible insulin secretion: special reference to expression of glucose transporter isoforms. Endocrinology 127(1), 126–132 (1990)CrossRefPubMed
47.
Zurück zum Zitat J.K. Ketterman, Y.V. Li, Presynaptic evidence for zinc release at the mossy fiber synapse of rat hippocampus. J. Neurosci. Res. 86(2), 422–434 (2008)CrossRefPubMed J.K. Ketterman, Y.V. Li, Presynaptic evidence for zinc release at the mossy fiber synapse of rat hippocampus. J. Neurosci. Res. 86(2), 422–434 (2008)CrossRefPubMed
48.
Zurück zum Zitat S.C. Burdette, G.K. Walkup, B. Spingler, R.Y. Tsien, S.J. Lippard, Fluorescent sensors for Zn(2+) based on a fluorescein platform: synthesis, properties and intracellular distribution. J. Am. Chem. Soc. 123(32), 7831–7841 (2001)CrossRefPubMed S.C. Burdette, G.K. Walkup, B. Spingler, R.Y. Tsien, S.J. Lippard, Fluorescent sensors for Zn(2+) based on a fluorescein platform: synthesis, properties and intracellular distribution. J. Am. Chem. Soc. 123(32), 7831–7841 (2001)CrossRefPubMed
49.
Zurück zum Zitat G.K. Walkup, S.C. Burdette, S.J. Lippard, R.Y. Tsien, A new cell-permeable fluorescent probe for Zn2+. J. Am. Chem. Soc. 122(23), 5644–5645 (2000). doi:10.1021/ja000868p CrossRef G.K. Walkup, S.C. Burdette, S.J. Lippard, R.Y. Tsien, A new cell-permeable fluorescent probe for Zn2+. J. Am. Chem. Soc. 122(23), 5644–5645 (2000). doi:10.​1021/​ja000868p CrossRef
50.
Zurück zum Zitat G. Anderegg, E. Hubmann, N.G. Podder, F. Wenk, Pyridinderivate als Komplexbildner. XI. Die Thermodynamik der Metallkomplexbildung mit Bis-, Tris- und Tetrakis[(2-pyridyl)methyl]-aminen. Helv. Chim. Acta 60(1), 123–140 (1977). doi:10.1002/hlca.19770600115 CrossRef G. Anderegg, E. Hubmann, N.G. Podder, F. Wenk, Pyridinderivate als Komplexbildner. XI. Die Thermodynamik der Metallkomplexbildung mit Bis-, Tris- und Tetrakis[(2-pyridyl)methyl]-aminen. Helv. Chim. Acta 60(1), 123–140 (1977). doi:10.​1002/​hlca.​19770600115 CrossRef
51.
Zurück zum Zitat P. Arslan, F. Di Virgilio, M. Beltrame, R.Y. Tsien, T. Pozzan, Cytosolic Ca2+ homeostasis in Ehrlich and Yoshida carcinomas. A new, membrane-permeant chelator of heavy metals reveals that these ascites tumor cell lines have normal cytosolic free Ca2+. J. Biol. Chem. 260(5), 2719–2727 (1985)PubMed P. Arslan, F. Di Virgilio, M. Beltrame, R.Y. Tsien, T. Pozzan, Cytosolic Ca2+ homeostasis in Ehrlich and Yoshida carcinomas. A new, membrane-permeant chelator of heavy metals reveals that these ascites tumor cell lines have normal cytosolic free Ca2+. J. Biol. Chem. 260(5), 2719–2727 (1985)PubMed
52.
Zurück zum Zitat K.Y. Chu, M.J. Briggs, T. Albrecht, P.F. Drain, J.D. Johnson, Differential regulation and localization of carboxypeptidase D and carboxypeptidase E in human and mouse beta-cells. Islets 3(4), 155–165 (2011)CrossRefPubMed K.Y. Chu, M.J. Briggs, T. Albrecht, P.F. Drain, J.D. Johnson, Differential regulation and localization of carboxypeptidase D and carboxypeptidase E in human and mouse beta-cells. Islets 3(4), 155–165 (2011)CrossRefPubMed
53.
Zurück zum Zitat S. Watkins, X. Geng, L. Li, G. Papworth, P.D. Robbins, P. Drain, Imaging secretory vesicles by fluorescent protein insertion in propeptide rather than mature secreted peptide. Traffic 3(7), 461–471 (2002)CrossRefPubMed S. Watkins, X. Geng, L. Li, G. Papworth, P.D. Robbins, P. Drain, Imaging secretory vesicles by fluorescent protein insertion in propeptide rather than mature secreted peptide. Traffic 3(7), 461–471 (2002)CrossRefPubMed
54.
Zurück zum Zitat D. Baetens, F. Malaisse-Lagae, A. Perrelet, L. Orci, Endocrine pancreas: three-dimensional reconstruction shows two types of islets of langerhans. Science 206(4424), 1323–1325 (1979)CrossRefPubMed D. Baetens, F. Malaisse-Lagae, A. Perrelet, L. Orci, Endocrine pancreas: three-dimensional reconstruction shows two types of islets of langerhans. Science 206(4424), 1323–1325 (1979)CrossRefPubMed
56.
Zurück zum Zitat D.M. Bers, C.W. Patton, R. Nuccitelli, A practical guide to the preparation of Ca2+ buffers. Methods Cell Biol. 40, 3–29 (1994)CrossRefPubMed D.M. Bers, C.W. Patton, R. Nuccitelli, A practical guide to the preparation of Ca2+ buffers. Methods Cell Biol. 40, 3–29 (1994)CrossRefPubMed
57.
Zurück zum Zitat Y. Li, C.J. Hough, C.J. Frederickson, J.M. Sarvey, Induction of mossy fiber –> Ca3 long-term potentiation requires translocation of synaptically released Zn2+. J. Neurosci. 21(20), 8015–8025 (2001)PubMed Y. Li, C.J. Hough, C.J. Frederickson, J.M. Sarvey, Induction of mossy fiber –> Ca3 long-term potentiation requires translocation of synaptically released Zn2+. J. Neurosci. 21(20), 8015–8025 (2001)PubMed
58.
Zurück zum Zitat H.L. Hellmich, C.J. Frederickson, D.S. DeWitt, R. Saban, M.O. Parsley, R. Stephenson, M. Velasco, T. Uchida, M. Shimamura, D.S. Prough, Protective effects of zinc chelation in traumatic brain injury correlate with upregulation of neuroprotective genes in rat brain. Neurosci. Lett. 355(3), 221–225 (2004)CrossRefPubMed H.L. Hellmich, C.J. Frederickson, D.S. DeWitt, R. Saban, M.O. Parsley, R. Stephenson, M. Velasco, T. Uchida, M. Shimamura, D.S. Prough, Protective effects of zinc chelation in traumatic brain injury correlate with upregulation of neuroprotective genes in rat brain. Neurosci. Lett. 355(3), 221–225 (2004)CrossRefPubMed
59.
Zurück zum Zitat T.J. Kamp, J.W. Hell, Regulation of cardiac L-type calcium channels by protein kinase A and protein kinase C. Circ. Res. 87(12), 1095–1102 (2000)CrossRefPubMed T.J. Kamp, J.W. Hell, Regulation of cardiac L-type calcium channels by protein kinase A and protein kinase C. Circ. Res. 87(12), 1095–1102 (2000)CrossRefPubMed
60.
Zurück zum Zitat G. Devis, G. Somers, E. Van Obberghen, W.J. Malaisse, Calcium antagonists and islet function. I. Inhibition of insulin release by verapamil. Diabetes 24(6), 247–251 (1975)CrossRefPubMed G. Devis, G. Somers, E. Van Obberghen, W.J. Malaisse, Calcium antagonists and islet function. I. Inhibition of insulin release by verapamil. Diabetes 24(6), 247–251 (1975)CrossRefPubMed
61.
Zurück zum Zitat S. Falkmer, R. Odselius, B. Blondel, M. Prentki, C.B. Wollheim, Energy dispersive X-ray microanalysis of zinc and calcium in organelles of insulin-producing cells of the mouse, rat, and a fish. Biomed. Biochim. Acta 44(1), 37–43 (1985)PubMed S. Falkmer, R. Odselius, B. Blondel, M. Prentki, C.B. Wollheim, Energy dispersive X-ray microanalysis of zinc and calcium in organelles of insulin-producing cells of the mouse, rat, and a fish. Biomed. Biochim. Acta 44(1), 37–43 (1985)PubMed
62.
Zurück zum Zitat U. Lindh, L. Juntti-Berggren, P.O. Berggren, B. Hellman, Proton microprobe analysis of pancreatic beta-cells. Biomed. Biochim. Acta 44(1), 55–61 (1985)PubMed U. Lindh, L. Juntti-Berggren, P.O. Berggren, B. Hellman, Proton microprobe analysis of pancreatic beta-cells. Biomed. Biochim. Acta 44(1), 55–61 (1985)PubMed
64.
Zurück zum Zitat W.J. Qian, C.A. Aspinwall, M.A. Battiste, R.T. Kennedy, Detection of secretion from single pancreatic beta-cells using extracellular fluorogenic reactions and confocal fluorescence microscopy. Anal. Chem. 72(4), 711–717 (2000)CrossRefPubMed W.J. Qian, C.A. Aspinwall, M.A. Battiste, R.T. Kennedy, Detection of secretion from single pancreatic beta-cells using extracellular fluorogenic reactions and confocal fluorescence microscopy. Anal. Chem. 72(4), 711–717 (2000)CrossRefPubMed
65.
Zurück zum Zitat D. Li, S. Chen, E.A. Bellomo, A.I. Tarasov, C. Kaut, G.A. Rutter, W.H. Li, Imaging dynamic insulin release using a fluorescent zinc indicator for monitoring induced exocytotic release (ZIMIR). Proc. Natl. Acad. Sci. U.S.A. 108(52), 21063–21068 (2011). doi:10.1073/pnas.1109773109 PubMedCentralCrossRefPubMed D. Li, S. Chen, E.A. Bellomo, A.I. Tarasov, C. Kaut, G.A. Rutter, W.H. Li, Imaging dynamic insulin release using a fluorescent zinc indicator for monitoring induced exocytotic release (ZIMIR). Proc. Natl. Acad. Sci. U.S.A. 108(52), 21063–21068 (2011). doi:10.​1073/​pnas.​1109773109 PubMedCentralCrossRefPubMed
68.
Zurück zum Zitat J.C. Henquin, M. Nenquin, P. Stiernet, B. Ahren, In vivo and in vitro glucose-induced biphasic insulin secretion in the mouse: pattern and role of cytoplasmic Ca2+ and amplification signals in beta-cells. Diabetes 55(2), 441–451 (2006)CrossRefPubMed J.C. Henquin, M. Nenquin, P. Stiernet, B. Ahren, In vivo and in vitro glucose-induced biphasic insulin secretion in the mouse: pattern and role of cytoplasmic Ca2+ and amplification signals in beta-cells. Diabetes 55(2), 441–451 (2006)CrossRefPubMed
70.
Zurück zum Zitat I.I. Nita, M. Hershfinkel, C. Kantor, G.A. Rutter, E.C. Lewis, I. Sekler, Pancreatic beta-cell Na+ channels control global Ca2+ signaling and oxidative metabolism by inducing Na+ and Ca2+ responses that are propagated into mitochondria. FASEB J. 28(8), 3301–3312 (2014). doi:10.1096/fj.13-248161 CrossRefPubMed I.I. Nita, M. Hershfinkel, C. Kantor, G.A. Rutter, E.C. Lewis, I. Sekler, Pancreatic beta-cell Na+ channels control global Ca2+ signaling and oxidative metabolism by inducing Na+ and Ca2+ responses that are propagated into mitochondria. FASEB J. 28(8), 3301–3312 (2014). doi:10.​1096/​fj.​13-248161 CrossRefPubMed
71.
Zurück zum Zitat L. Aguilar-Bryan, J. Bryan, Molecular biology of adenosine triphosphate-sensitive potassium channels. Endocr. Rev. 20(2), 101–135 (1999)PubMed L. Aguilar-Bryan, J. Bryan, Molecular biology of adenosine triphosphate-sensitive potassium channels. Endocr. Rev. 20(2), 101–135 (1999)PubMed
74.
Zurück zum Zitat M. Hutton, The effects of environmental lead exposure and in vitro zinc on tissue delta-aminolevulinic acid dehydratase in urban pigeons. Comp. Biochem. Physiol. C 74(2), 441–446 (1983)CrossRefPubMed M. Hutton, The effects of environmental lead exposure and in vitro zinc on tissue delta-aminolevulinic acid dehydratase in urban pigeons. Comp. Biochem. Physiol. C 74(2), 441–446 (1983)CrossRefPubMed
76.
Zurück zum Zitat F. Chimienti, S. Devergnas, A. Favier, M. Seve, Identification and cloning of a beta-cell-specific zinc transporter, ZnT-8, localized into insulin secretory granules. Diabetes 53(9), 2330–2337 (2004)CrossRefPubMed F. Chimienti, S. Devergnas, A. Favier, M. Seve, Identification and cloning of a beta-cell-specific zinc transporter, ZnT-8, localized into insulin secretory granules. Diabetes 53(9), 2330–2337 (2004)CrossRefPubMed
77.
Zurück zum Zitat N. Wijesekara, F.F. Dai, A.B. Hardy, P.R. Giglou, A. Bhattacharjee, V. Koshkin, F. Chimienti, H.Y. Gaisano, G.A. Rutter, M.B. Wheeler, Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia 53(8), 1656–1668 (2010). doi:10.1007/s00125-010-1733-9 CrossRefPubMed N. Wijesekara, F.F. Dai, A.B. Hardy, P.R. Giglou, A. Bhattacharjee, V. Koshkin, F. Chimienti, H.Y. Gaisano, G.A. Rutter, M.B. Wheeler, Beta cell-specific Znt8 deletion in mice causes marked defects in insulin processing, crystallisation and secretion. Diabetologia 53(8), 1656–1668 (2010). doi:10.​1007/​s00125-010-1733-9 CrossRefPubMed
78.
Zurück zum Zitat T.J. Nicolson, E.A. Bellomo, N. Wijesekara, M.K. Loder, J.M. Baldwin, A.V. Gyulkhandanyan, V. Koshkin, A.I. Tarasov, R. Carzaniga, K. Kronenberger, T.K. Taneja, G. da Silva Xavier, S. Libert, P. Froguel, R. Scharfmann, V. Stetsyuk, P. Ravassard, H. Parker, F.M. Gribble, F. Reimann, R. Sladek, S.J. Hughes, P.R. Johnson, M. Masseboeuf, R. Burcelin, S.A. Baldwin, M. Liu, R. Lara-Lemus, P. Arvan, F.C. Schuit, M.B. Wheeler, F. Chimienti, G.A. Rutter, Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetes-associated variants. Diabetes 58(9), 2070–2083 (2009). doi:10.2337/db09-0551 PubMedCentralCrossRefPubMed T.J. Nicolson, E.A. Bellomo, N. Wijesekara, M.K. Loder, J.M. Baldwin, A.V. Gyulkhandanyan, V. Koshkin, A.I. Tarasov, R. Carzaniga, K. Kronenberger, T.K. Taneja, G. da Silva Xavier, S. Libert, P. Froguel, R. Scharfmann, V. Stetsyuk, P. Ravassard, H. Parker, F.M. Gribble, F. Reimann, R. Sladek, S.J. Hughes, P.R. Johnson, M. Masseboeuf, R. Burcelin, S.A. Baldwin, M. Liu, R. Lara-Lemus, P. Arvan, F.C. Schuit, M.B. Wheeler, F. Chimienti, G.A. Rutter, Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetes-associated variants. Diabetes 58(9), 2070–2083 (2009). doi:10.​2337/​db09-0551 PubMedCentralCrossRefPubMed
Metadaten
Titel
Autocrine effect of Zn2+ on the glucose-stimulated insulin secretion
verfasst von
Kira G. Slepchenko
Nigel A. Daniels
Aili Guo
Yang V. Li
Publikationsdatum
01.09.2015
Verlag
Springer US
Erschienen in
Endocrine / Ausgabe 1/2015
Print ISSN: 1355-008X
Elektronische ISSN: 1559-0100
DOI
https://doi.org/10.1007/s12020-015-0568-z

Weitere Artikel der Ausgabe 1/2015

Endocrine 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.